Int J App Pharm, Vol 11, Issue 6, 2019, 12-17Review Article


HUMAN ORGANIC SOLUTE TRANSPORTERS UTTERED IN SMALL INTESTINE, LIVER, AND KIDNEY FOR HOMEOSTASIS

HARANATH CHINTHAGINJALA, HINDUSTAN ABDUL AHAD*, MAHESH REDDY CHALLA, GANDLA CHAITHANYA BARGHAV, PASAM DEVIKA, SRIKANTHAM SAI VIKAS

Department of Pharmaceutics and Industrial Pharmacy, Raghavendra Institute of Pharmaceutical Education and Research (RIPER)-Autonomous, K. R. Palli Cross, Ananthapuramu 515721, Andhra Pradesh, India
Email: abdulhindustan@gmail.com

Received: 06 Aug 2019, Revised and Accepted: 05 Oct 2019


ABSTRACT

The transporters participate in a significant role in drug absorption, distribution, metabolism, and elimination. Transporters are of efflux and influx type, need ATP-binding sites for their in and out movement across the cell membrane. These transporters play an important role in allowing or opposing the drugs into the cells, results in non-linearity in drug pharmacokinetics. A wide range of transporters was discovered; among them, organic solute transporters (OST) play a key role in drug absorption and disposition. Organic solute transporters is a heteromeric transporter localized to the basolateral of epithelial cells. It is the primary efflux bile acid transporter in the intestine of mammals.

Keywords: Organic solute transporters, ATP-binding site, Bile acid transporter


INTRODUCTION

A drug transporter (TRS) plays a dynamic role in pharmacokinetics (PK) of an orally given drug. The drug TRS at the GIT and the liver govern the passage of drugs into the systemic flow. The Biopharmaceutics Drug Distribution and Classification System (BDDCS) is a beneficial tool in forecasting the effects that a drug TRS in the GIT and the liver, which have an impact on the drug’s PK. The BDDCS is anticipated by Amidon et al., which is the Biopharmaceutics Classification System (BCS) modifications based on drug penetrability and solubility [1].

A lot of work has distinguished and described on intestinal and hepatic TRS concerning tissue articulation profiles, guidelines, and instruments of transport, species contrasts, and hereditary polymorphisms. Both influx and efflux TRS (ET) is vital in deciding oral medication demeanor by controlling retention and bioavailability (BA). This significant influx and ET in charge of xenobiotic transport have a place with the two superfamilies namely Solute Carrier (SLC), and Solute Carrier Organic anion (SLCO) group. The SLC superfamily transport Type I natural anions, cations, and zwitterions, whereas the SLCO superfamily transports Type II natural anions. The SLC and SLCO TRS are of Uniporter, Antiporteror Symporter type, not all the TRS have been completely explained.

The SLC superfamily envelops an assortment of TRS, including the Organic Anion TRS(OAT), the Organic Cation TRS (OCT), the Organic zwitterions/Cation TRS (OCTN), the Equilibrate Nucleoside TRS (ENT), the Concentrative Nucleoside TRS (CNT), the Apical Sodium dependent bile salt TRS (ASBT), the Mono Carboxylate TRS (MCT), and the Peptide TRS (PEPT) [2]. The SLCO family comprises the Organic Anion Transporting Polypeptides (OATP). ET communicated in the digestive tract and the liver incorporate P-glycoprotein (Pgp), Bile Salt Export Pump (BSEP), the Multidrug Resistant Protein(MRP), and Breast Cancer Resistant Protein (BCRP). All these TRS need ATP as a vitality source, enabling them to move against the concentration gradient. These TRS appearances in the gut, liver, BBB and, renal tubules influence the movement of drugs and, vital role in determining oral medication PK [3]. The major human SLC drug TRS uttered in the small intestine, liver, and kidney were shown in table 1.

TRS for intestinal drug absorption

The integration of medications from the gut is a basic factor in deciding oral BA. Enterocytes of the small intestine (fig. 1) is outfitted with a variety of deluge TRS at the luminal film for the consumption of sustenance parts and medications. Various SLC mediates transporting proteins have been portrayed at the brush outskirt layer of human enterocytes, including PEPT1, OATP1A2, OATP2B1, OCTN1, and OCTN2 [23]. Articulation dimensions of a slice of these TRS seem to change along the gut, however, results from various examinations on mRNA and protein articulation don't harmony, aside from PEPT1, which is dominatingly communicated in the little. PEPT1 perceives different peptide-like medications and focusing on this TRS has been utilized to improve the oral BA of inadequately ingested medications (E. g., Nucleoside analogs). The Influx of cationic medications from the gut is intervened by OCTN1 and OCTN2 [24], which is invigorated by electro impartial cation-cation exchange. Changes in the qualities encoding for these TRS have been related to incendiary gut malady and polymorphisms could be of effect on cationic medication retention [25]. OATP1A2 and OATP2B1 are in charge of the take-up of an expansive scope of amphipathic drugs. While there is very some cover in particularity, a few substrates are specially or solely transported by one of them. For instance, just OATP1A2 [26] can intervene fexofenadine take-up and the presumable focus of restraint by grapefruit squeeze. The initial phase in the discharge of cationic medications from blood to gut lumen is interceded by OCT1 in the basolateral film, trailed by the activity of ET in the brush fringe layer. These are the OCTNs that can likewise work as secretory TRS by trading luminal natural cations against a higher convergence of intracellular cationic medications. Also, MDR1/P-gp siphons decidedly charged hydrophobic medications again into the lumen, which could have entered the cells by uninvolved dispersion. The ABC TRS (ATP-binding cassette TRS) P-gp, MRP2, and BCRP are all articulated in the brush border membrane where they have a significant role as the porter in the gut, restrictive the oral BA of many drugs. The inflection of their activity with selective inhibitors could be a useful strategy to increase the oral BA of drugs [27].

TRS for hepatic drug elimination

The liver has a striking capacity to productively extricate drugs with high protein officials from the blood course. The hepatic take-up of medications is much of the time pursued by Phase I and Phase II biotransformation and efflux of the metabolites into bile and adds to the hepatic first-pass impact. The Influx and ET communicated at the basolateral and apical layer of the hepatocytes have been perceived as basic determinants in medication disposal Drug convergence TRS communicated at the sinusoidal film incorporates OATP1B1, OATP1B3, OATP2B1, OAT2, and OCT1. Specifically, OATP1B1 is perceived as a critical take-up TRS for some clinically applicable medications, for example, macrolide anti-infection agents, statins (HMG-CoA reductase inhibitors), Glitazones (Thiazolidinediones), Sartans (angiotensin II receptor adversaries), and angiotensin-changing over chemical (ACE) inhibitors [28].

Table 1: Major human SLC drug TRS uttered in the small intestine, liver, and kidney

Protein Mechanism

Tissue

distribution

Examples of drug substrates References
PEPT1

H+/peptide

symporter

Intestine

Kidney

Amoxicillin, Ampicillin, Bestatin, Cefaclor, Cefadroxil, Cefixime, Enalapril, Midodrine, Temocapril, and Temocaprilat [4]
PEPT2

H+/peptide

symporter

Kidney Amoxicillin, Bestatin, Cefaclor, Cefadroxil, and Valganciclovir [5]
OCT1 OC uniporter

Intestine

Liver

Acyclovir, Cimetidine, Ganciclovir, Metformin, Quinine, Quinidine, and Zidovudine [6]
OCT2 OC uniporter Kidney Cimetidine, Cisplati, Famotidine, Mepiperphenidol, Memantine, Ranitidine, Metformin, Propranolol, Pancuronium, Quinine, and Zidovudine, [7]
OCTN1

H+or OC

antiporter

Intestine

Kidney

Ergothioneine, Gabapentin, Mepyramine, Quinidine, and Verapamil [9]
OCTN2

OC antiporter

Na+symporter

(carnitine)

Intestine

Kidney

Cephaloridine, Emetine, Mepyramine, Quinidine, Verapamil, and Valproate [8]
OAT1

DC/OA

antiporter

Kidney Acyclovir, Adefovir, Bumetanide, Cimetidine, Cidofovir, Didanosine, Ganciclovir, Furosemide, Ibuprofen, Indomethacin, Ketoprofen, Methotrexate, PAH, Stavudine, Trifluridine, Tetracycline, Tenofovir, Zalcitabine and Zidovudine [11]
OAT2 OA antiporter Liver 5-Fluorouracil, Allopurinol, Bumetanide, Cimetidine, Erythromycin, Methotrexate, PAH, Ranitidine, Taxol, Salicylate, Theophylline, and Zidovudine [12]
OAT3

DC/OA

antiporter

Kidney Benzyl Penicillin, Cimetidine, Furosemide, Ibuprofen, Indomethacin, Ketoprofen, Methotrexate, Olmesartan, PAH, Pravastatin, Ranitidine, Salicylate, Tetracycline, Valacyclovir, Zidovudine [13]
OAT4

Cl–/OA

antiporter

Kidney Bumetanide, Ketoprofen, Methotrexate, PAH, Salicylate, Tetracycline, and Zidovudine [14]
OATP1A2 OA antiporter Kidney(DT), Intestine Enalapril, Fexofenadine, Indomethacin, Levofloxacin, Ouabain, Rosuvastatin, Rocuronium, Pitavastatin, and Temocaprilat [15]
OATP1B1 OA antiporter Liver Atorvastatin, Bosentan, Benzyl Penicillin, Caspofungin, Cerivastatin, Enalapril, Fexofenadine, Fluvastatin, Olmesartan, Pravastatin, Pitavastatin, Rosuvastatin, Rifampicin, Simvastatin, Troglitazone, and Valsartan [16]
OATP1B3 OA antiporter Liver Bosentan, Digoxin, Enalapril, Fluvastatin Fexofenadine, Methotrexate, Ouabain, Paclitaxel, Pitavastatin, Rifampicin, Rosuvastatin, Telmisartan, and Valsartan [17]
OATP2B1 OA antiporter

Liver

Intestine

Atorvastatin, Benzylpenicillin, Bosentan, Fluvastatin, Glibenclamide, Pravastatin, Pitavastatin, and Rosuvastatin [18]

MDR1/

P-gp

Primary

active

Intestine

Liver

Kidney

Amiodarone, Bisantrene, Carbamazepine, Celiprolol, Chloroquine, Colchicine, Cyclosporin-A, Daunorubicin, Desipramine, Digitoxin, Digoxin, Docetaxel, Doxorubicin, Erythromycin, Etoposide, Fexofenadine, Grepafloxacin, Imatinib, Indinavir, Ivermectin, Levofloxacin, Lidocaine, Loperamide, Losartan, Lovastatin, Methadone, Methotrexate, Mibefradil, Mitoxantrone, Morphine, Nelfinavir, Ortataxel, Paclitaxel, Paclitaxel, Ritonavir, Saquinavir, Sirolimus, Sparfloxacin, Sumatriptan, Tacrolimus, Talinolol, Terfenadine, Topotecan, Vecuronium, Vinblastine, and Vincristine [19]
MRP2

Primary

active

Intestine

Liver

Kidney

Cisplatin, Doxorubicin, Etoposide, Grepafloxacin, Glutathione conjugates, Indinavir, Methotrexate, Ritonavir, Saquinavir, Vinblastine, and Vincristine [20]
MRP3

Primary

active

Intestine

Liver

Kidney

Acetaminophen, Glucuronide conjugates, Ethinyl estradiol, Etoposide, Morphine and Methotrexate
MRP4

Primary

active

Intestine

Liver

Kidney

6-Mercaptopurine, 6-Thioguanine, Adefovir, Ceftizoxime, Cefazolin, Cefotaxime, Cefmetazole, Edaravone, Furosemide, Glucuronide, Hydrochlorothiazide, Leucovorin, Methotrexate, Olmesartan, PAH, Topotecan, and Tenofovir [12]
BCRP

Primary

active

Intestine

Liver

Kidney

Abacavir, Albendazole, Camptothecin, Cerivastatin, Cimetidine, Ciprofloxacin, Dipyridamole, Edaravone, Erlotinib, Flavopiridol, Glibenclamide, Gefitinib, Imatinib, Lamivudine, Methotrexate, Mitoxantrone, Nelfinavir, Nitrofurantoin, Norfloxacin, Ofloxacin, Oxfendazole, Pitavastatin, Rosuvastatin, Olmesartan, Sulfasalazine, Sulfoxide, Topotecan, and Zidovudine [22]

TRS for renal drug elimination

The renal treatment of medications includes aloof procedures, including glomerular filtration and back dissemination along the nephron, and bearer intervened emission and re-absorption that are for the most part situated in the proximal tubule. For most medications that experience bearer interceded transport in the kidney (fig. 2), renal emission can be considered as a vectorial procedure, including the take-up of substances from the blood over the basolateral film of proximal rounded cells, trailed by their efflux over the brush outskirt layer the pee. At the basolateral film, separate inundation TRS exists for the take-up of principally type-I natural anions and cations, which are prominent for their high leeway limit, a wide assortment of substrates acknowledged, and inclusion in medication tranquilize associations.

As a result of proficient take-up, numerous medications will in general amass in the cell here and there, causing nephrotoxicity [29].

Fig. 1: Diagram representing major drug TRS in enterocytes of the human small intestine

Fig. 2: Schematic model of the major drug TRS in human renal proximal tubular cells

The take-up of anionic medications at the basolateral layer of the renal proximal tubule is controlled by OAT1 and OAT3. The two TRS have covering substrate specificities and offer a similar method of transport driven by the trading of natural anions with dicarboxylates. OAT1 has a higher fondness for hydrophilic natural anions with little atomic loads (type I), like PAH, Adefovir, cidofovir, and Tenofovir. OAT3 likewise transports some amphipathic natural anions (type II) that are liver OATP substrates, including Benzyl Penicillin, Pravastatin, Olmesartan, and even some cationic medications (E. g., Cimetidine, and Ranitidine). The more extensive explicitness, just as the generally higher renal articulation dimensions of OAT3 contrasted with OAT1, recommends an increasingly articulated job of OAT3 in human renal natural anion transport. Serious medication sedate communications have been accounted for among methotrexate and NSAIDs because of rivalry for OAT1-and OAT3-intervened take-up, although the cooperation at the dimension of the apical ET viz., MRP2 and MRP4 most likely additionally adds to this component.

Organic solute TRS

Natural solute TRS alpha-beta (OSTα-Ostβ) is a heteromeric TRS confined to the basolateral film of epithelial cells associated with sterol transport [30]. It is accepted to be the essential bile corrosive ET in the digestive tract, of next to these lines fundamental to bile corrosive homeostasis and the enterohepatic course. It can transport an assortment of bile acids, just as estrone 3-sulfate, dehydroepiandrosterone 3-sulfate, digoxin, and prostaglandin E2 [31].

Articulation of the two subunits is variable among species and tissues; in people, high articulation is noted in the liver, small digestive tract, kidney, testis, and adrenal organ. OSTα-Ostβ is straightforwardly controlled by the bile corrosive detecting atomic receptor, farnesoid X receptor (FXR) [32].

Moreover, it is a piece of the complex administrative pathway that controls bile corrosive amalgamation and homeostasis. Hepatic OSTα-Ostβ is up delimited in cholestasis in the two people and rodents, where it seems to assume a defensive job. Extra investigations are important to decide its job in liver damage, bile corrosive malabsorption, and lipid and glucose digestion, just as a potential defensive job for kidney OSTα-Ostβ in cholestasis [33]. The structure of the OST transporter illustrated in fig. 3.

Fig. 3: Structure of OST-TRS

Characteristics of Ostα-Ostβ

Ostα encodes for a protein of 352 amino acids and seven putative transmembrane spaces, though skate Ostβ is anticipated to contain 182 amino acids and is a solitary film spreading over protein [34]. Human OSTα and mouse Ostα share 83% amino corrosive personality with one another and 41% amino corrosive character with skate OSTα. Human OSTβ shares 63% amino corrosive personality with mouse OSTβ and just 25% amino corrosive character with skate Ostβ [35]. Articulation of the two subunits is required for transport and mammalian orthologues can practically supplement the first skate proteins regardless of the distinctions in corrosive amino homology. Human OSTα-OSTβ can transport estrone 3-sulfate, digoxin, and prostaglandin E2, just as taurocholate, yet not estradiol 17β-D-glucuronide or paminohippurate [36].

Analysis of communicated grouping label includes in people has affirmed that OSTα and OSTβ are almost bottomless in the steroid rich organs, for example, liver, digestive tract, kidney, testis, mammary organ, uterus, prostate, and thyroid. In mice and rodents, the articulation is most elevated in the small intestinal system and the kidney, where its dispersion mirrors that of the corrosive bile take-up TRS [37] have proposed that rodents may have a higher intestinal articulation of OSTα-Ostβ because they require a higher rate of dietary sterol assimilation than people. Strangely, the small intestine does not seem to show the noticeable angle of appropriation from the duodenum to ileum that has been found in the mouse and this might be because of contrasts in the pool of conjugated bile corrosive among human and mouse. In endocrine organs, it might capacity to exchange steroid hormones among tissues and blood [38].

Regulation of Ostα-Ostβ

A standout amongst the most imperative parts of OSTα-OSTβ is its capacity to be emphatically controlled by bile acids through the atomic receptor, Farnesoid X receptor This atomic receptor controls bile corrosive homeostasis by keeping up a fine equalization in bile corrosive blend and transport by directing key qualities in the liver, kidney and gastric system [39].

In this way, bile acids can curb their very own blend through authoritative to FXR in the liver and digestive tract and invigorating translation of short heterodimer accomplice (SHP) and fibroblast development factor 19 (FGF19), and hindering CYP7A1, CYP8B1 and liver receptor homolog 1 (LRH-1).17 [40].

In the human, two putative IR-1/FXREs have been recognized in the OSTα advertiser and one in the OSTβ promoter. The mRNA and protein articulation of the two subunits can be incited by the FXR agonists’ Chenodeoxycholic corrosive (CDCA) and GW4064, in different human cell lines, including the hepatocyte lines, HepG2, and Huh7. Reduction of FXR by with FXR-explicit siRNAs annulled this agonist-initiated OSTα-OSTβ articulation in Huh7 cells.

In the mouse [41], one potential Fare grouping has been accounted for in both the Ostα and Ostβ promoters.15 Basal dimensions of Ostα and Ostβ are lower in ileum from Fxr mice and GW4064 treatment of organ culture of adrenal organs from these mice neglect to actuate Ostα or Ostβ.[42]

OSTα-OSTβ TRS activity

The system for OSTα-OSTβ intervened transport has not been completely clarified. OSTα-OSTβ intervened transport was unaffected by consumption of intracellular ATP, by adjustments in Trans layer electrolyte focus slopes, or by changes in the pH inclination. OSTα-OSTβ displays both take-up and efflux properties, and solute transport are trans-invigorated by known substrates.

The OSTα-OSTβ works by encouraging dissemination and intercedes solute take-up or efflux, contingent upon the solute's electrochemical angle. A methodical screening exertion to recognize OSTα-OSTβ transport substrates or watchful correlation of the substrate particularity of OST from various species has not yet been distributed and this zone remains generally unexplored. OSTα-OSTβ substrates incorporate the real types of bile acids, including glycine and taurine conjugates of Cholic Corrosive, Deoxycholic Corrosive, Chenodeoxycholic Corrosive, and Ursodeoxycholic Corrosive, just as non-bile corrosive substrates.

E. g., Estrone-3-sulfate, Digoxin, Prostaglandin E2, and Dehydroepiandrosterone-3-sulfate (DHEAS) [43].

E. g., Spironolactone, Bromo sulfophthalein, Probenecid, and indomethacin. These primer outcomes recommend that the substrate particularity for OSTα-OSTβ is generally wide and is predictable with an immediate job in the vehicle of different solutes as well as steroids or steroid sulfates [44].

Other clinical implications of OSTα-OSTβ

In any case, given its significance in enterohepatic flow and bile corrosive homeostasis, just as intestinal lipid ingestion, extra investigations will be expected to take a gander at its job in bile corrosive poor absorption, crabby inside disorders, enterocolitis, cholelithiasis, and lipid and glucose digestion. An ongoing report researched the job of different ileal bile corrosive TRS in essential idiopathic bile corrosive poor absorption (IBAM), which might be in charge of 30–half of the patients with unexplained endless diarrhea [45].

In any case, OSTα-OSTβ in this disease presently can't seem to be analyzed. Necrotizing enter colitis is another serious the runs ailment seen generally in premature neonates where the anomalous aggregation of bile acids in the distal small digestive tract may assume a job in its pathogenesis [46].

Articulation of OSTα-OSTβ has likewise been analyzed in fat and non-fat patients with gallstone illness. A noteworthy decrease in both mRNA and protein articulation of both OSTα and OSTβ was found in ordinary weight gallstone TRS, however not in controls or corpulent gallstone bearers. These progressions corresponded decidedly with the declaration of ASBT, IBABP, and FXR, proposing a job for these proteins in gallstone ailment in non-stout patients [47].

CONCLUSION

Ostα-Ostβ seems to have a vital role in protecting the ileal epithelium towards bile acid buildup and injury. Inactivation of Ostα resulted in the augmented countenance of FXR target genes as well as villous blunting, cell apoptosis, and oxidative stress in early perinatal growth. Ostα-Ostβ is a major mechanism for intestinal basolateral bile acid transport. Unlike blocking apical bile acid transport, blocking basolateral bile acid transport results in reduced hepatic bile acid synthesis, even in the face of a markedly reduced bile acid pool size. Whereas inhibiting Ostα-Ostβ could potentially raise plasma cholesterol levels by decreasing the hepatic conversion of cholesterol to bile acids, the combination of reduced return of bile acids in the enterohepatic circulation and decreased hepatic bile acid synthesis might be exploited therapeutically to relieve the hepatic bile acid burden in some forms of cholestatic liver disease. Thus, the stable association of both subunits may be required for Transporters function, or the Ost subunit may function as a chaperone to promote the egress of Ost and possibly other proteins from the endoplasmic reticulum., the mechanism by which these two proteins interact, their roles in generating a useful complex at the plasma membrane, and their roles in solute transport.

ACKNOWLEDGMENT

We are thankful to Prof. Padmanabha Reddy, Principal, RIPER, Ananthapuramu, AP, India for encouraging for doing this work.

CONFLICT OF INTERESTS

No conflict of interest was declared by the authors.

REFERENCES

  1. Amidon GL, Lennernas H, Shah VP, Crison JR. A theoretical basis for a biopharmaceutic drug classification: the correlation of in vitro drug product dissolution and in vivo bioavailability. Pharm Res 1995;12:413-20.

  2. Hertanto R, Bastian WP, Louisa M. The modulation of drug efflux transporter by curcumin in MCF 7 breast cancer cells after repeated exposure of endoxifen and estradiol. Int J Appl Pharm 2018;10:102-5.

  3. Dawson PA, Hubbert ML, Rao A. Getting the mOST from OST: role of organic solute transporters, OSTα-OSTβ, in bile acid and steroid metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2010;1801:994-1004.

  4. Russel FG. Transporters: importance in drug absorption, distribution, and removal. In: Enzyme-and transporters-based drug-drug interactions; 2010. p. 27-49.

  5. Liang Y, Li S, Chen L. The physiological role of drug transporters. Protein Cell 2015;6:334-50.

  6. Petzinger E, Geyer J. Drug transporters in pharmacokinetics. Naunyn-Schmiedeberg's Arch Pharmacol 2006;372:465-75.

  7. Ballatori N. Biology of a novel organic solute and steroid transporters, OSTα-OSTβ. Exp Biol Med 2005;230:689-98.

  8. Ballatori N, Christian WV, Wheeler SG, Hammond CL. The heteromeric organic solute transporters, OSTα–OSTβ/SLC51: a transporter for steroid-derived molecules. Mol Aspects Med 2013;34:683-92.

  9. Dawson PA, Hubbert ML, Rao A. Getting the mOST from OST: role of organic solute transporters, OSTα-OSTβ, in bile acid and steroid metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2010;1801:994-1004.

  10. Dawson PA, Lan T, Rao A. Bile acid transporters. J Lipid Res 2009;50:2340-57.

  11. Schaffner CA, Mwinyi J, Gai Z, Thasler WE, Eloranta JJ, Kullak Ublick GA. The organic solute transporters alpha and beta are induced by hypoxia in human hepatocytes. Liver Int 2015;35:1152-61.

  12. Rao A, Haywood J, Craddock AL, Belinsky MG, Kruh GD, Dawson PA. The organic solute transporters α-β, Ostα-Ostβ, is essential for intestinal bile acid transport and homeostasis. Proc Natl Acad Sci India 2008;105:3891-6.

  13. Anusha GB, Hindustan AA, Haranath C, Suryaprakash RC, Kalpana K. A technical view on transporters-the drug pharmacokinetics dictators. MOJ Bioequivalence Bioavailability 2019;6:39‒45.

  14. Landowski CP, Sun D, Foster DR, Menon SS, Barnett JL, Welage LS. Gene expression in the human intestine and correlation with oral valacyclovir pharmacokinetic parameters. J Pharmacol Exp Ther 2003;306:778-86.

  15. Shugarts S, Benet LZ. The role of transporters in the pharmacokinetics of orally administered drugs. Pharm Res 2009;26:2039-54.

  16. Mizuno N, Sugiyama Y. Drug transporters: their role and importance in the selection and development of new drugs. Drug Metab Pharmacokinet 2002;17:93-108.

  17. Wu CY, Benet LZ. Predicting drug disposition via application of BCS: transport/absorption/elimination interplay and development of a biopharmaceutics drug disposition classification system. Pharm Res 2005;22:11-23.

  18. Hagenbuch B, Gui C. Xenobiotic transporters of the human organic anion transporting polypeptides (OATP) family. Xenobiotica 2008;38:778-801.

  19. Zhou SF, Wang LL, Di YM, Xue CC, Duan W, Li CG. Substrates and inhibitors of human multidrug resistance-associated proteins and the implications in drug development. Curr Med Chem 2008;15:1981-2039.

  20. Nielsen S, Westerhoff AM, Ge LG, Carlsen KL, Pedersen MDL, Nielsen CU. MRP2-mediated transport of etoposide in MDCKII MRP2 cells is unaffected by commonly used non-ionic surfactants. Int J Pharm 2019;565:306-15.

  21. Kusuhara H, Sugiyama Y. ATP-binding cassette, subfamily G (ABCG family). Pflugers Archiv Eur J Physiol 2007;453:735-44.

  22. Srimaroeng C, Perry JL, Pritchard JB. Physiology, structure, and regulation of the cloned organic anion transporters. Xenobiotica 2008;38:889-935.

  23. Zair ZM, Eloranta JJ, Stieger B, Kullak Ublick GA. Pharmacogenetics of OATP (SLC21/SLCO), OAT and OCT (SLC22) and PEPT (SLC15) transporters in the intestine, liver and kidney. Pharmacogenomics 2008;9:597-624.

  24. Ciarimboli G. Organic cation transporters. Xenobiotica 2008;38:936-71.

  25. Van Montfoort JE, Muller M, Groothuis GM, Meijer DK, Koepsell H, Meier PJ. Comparison of “type I” and “type II” organic cation transport by organic cation Transporters and organic anion-transporting polypeptides. J Pharmacol Exp Ther 2001;298:110-5.

  26. Li L, Meier PJ, Ballatori N. Oatp2 mediates bidirectional organic solute transport: a role for intracellular glutathione. Mol Pharmacol 2000;58:335-40.

  27. Satlin LM, Amin V, Wolkoff AW. Organic anion transporting polypeptide mediates organic anion/HCO3− exchange. J Biol Chem 1997;272:26340-5.

  28. Rao A, Haywood J, Craddock AL, Belinsky MG, Kruh GD, Dawson PA. The organic solute transporters α-β, Ostα-Ostβ, is essential for intestinal bile acid transport and homeostasis. Proc Natl Acad Sci USA 2008;105:3891-6.

  29. Soroka CJ, Mennone A, Hagey LR, Ballatori N, Boyer JL. Mouse organic solute Transporters α deficiency enhances renal excretion of bile acids and attenuates cholestasis. Hepatology 2010;51:181-90.

  30. Mahadevan MV, Ramaswamy RS, Banumathi V. Mimosa pudica exerts neuroprotection against MPP induced neurotoxicity in SH-SY5Y cell lines-an in vitro model of anti-parkinsonism. Int J Pharm Pharm Sci 2006;9:21-6.

  31. Li N, Cui Z, Fang F, Lee JY, Ballatori N. Heterodimerization, trafficking and membrane topology of the two proteins, Ostα and Ostβ, that constitute the organic solute and steroid transporters. Biochem J 2007;407:363-72.

  32. Frankenberg T, Rao A, Chen F, Haywood J, Shneider BL, Dawson PA. Regulation of the mouse organic solute transporters α-β, Ostα-Ostβ, by bile acids. Am J Physiol Gastrointest Liver Physiol 2006;290:G912-22.

  33. Landrier JF, Eloranta JJ, Vavricka SR, Kullak Ublick GA. The nuclear receptor for bile acids, FXR, transactivates the human organic solute transporters-α and-β genes. Am J Physiol Gastrointest Liver Physiol 2006;290:G476-85.

  34. Sultan M, Rao A, Elpeleg O, Vaz FM, Abu Libdeh B, Karpen SJ. Organic solute transporters‐β (SLC51B) deficiency in two brothers with congenital diarrhea and features of cholestasis. Hepatology 2018;68:590-8.

  35. Khandelwal VK, Singh P, Ravingerova T, Chaudhary M. Evaluation of ezenus in an experimental model of diet-induced alcoholic and non-alcoholic fatty liver condition in rats. Int J Pharm Pharm Sci 2015;7:247-52.

  36. Russell DW. Fifty years of advances in bile acid synthesis and metabolism. J Lipid Res 2009;50(Suppl):S120-5.

  37. Malinen MM, Ali L, Bezençon J, Beaudoin JJ, Brouwer KLR. Organic solute Transporters OSTα/β is overexpressed in nonalcoholic steatohepatitis and modulated by drugs associated with liver injury. Am J Physiol Gastrointest Liver Physiol 2018;314:G597-G609.

  38. Courtney BF, Jianing Li, Jamie H, Kimberly P, Brian SR, Benjamin H. Organic solute transporters α-β protects ileal enterocytes from bile acid-induced injury. Cellular Mol Gastro Enterol Hepatol 2018;5:499–522.

  39. Khan AA, Chow EC, Porte RJ, Pang KS, Groothuis GM. Expression and regulation of the bile acid transporters, OSTα‐OSTβ in rat and human intestine and liver. Biopharm Drug Dispos 2009;30:241-58.

  40. Sun AQ, Balasubramaniyan N, Xu K, Liu C, Ponamgi VM, Liu H, et al. Protein-protein interaction and membrane localization of human organic solute transporters (hOST). Am J Physiol Gastrointest Liver Physiol 2007;292:G1586–93.

  41. Li N, Cui Z, Fang F, Lee JY, Ballatori N. Heterodimerization, trafficking and membrane topology of the two proteins, Ostα and Ostβ, that constitute the organic solute and steroid transporters. Biochem J 2007;407:363-72.

  42. Ballatori N, Christian WV, Lee JY, Dawson PA, Soroka CJ, Boyer JL. OSTα‐OSTβ: A major basolateral bile acid and steroid transporters in human intestinal, renal, and biliary epithelia. Hepatology 2005;42:1270-9.

  43. Melina MM, Izna Ali, Kim LR. Organic solute transporters OSTα/β is verexpressed in nonalcoholic steatohepatitis and modulated by drugs associated with liver injury. Am J Physiol Gastrointest Liver Physiol 2018;314:G597–G609.

  44. Seward DJ, Koh AS, Boyer JL, Ballatori N. Functional complementation between a novel mammalian polygenic transport complex and evolutionarily ancient organic solute transporters, OSTα-OSTβ. J Biol Chem 2003;278:27473-82.

  45. Dawson PA, Haywood J, Craddock AL, Wilson M, Tietjen M, Kluckman K. Targeted deletion of the ileal bile acid transporters eliminates enterohepatic cycling of bile acids in mice. J Biol Chem 2003;278:33920-7.

  46. Soroka CJ, Mennone A, Hagey LR, Ballatori N, Boyer JL. Mouse organic solute Transporters α deficiency enhances renal excretion of bile acids and attenuates cholestasis. Hepatology 2010;51:181-90.

  47. Al-mamun MA, Rahman MA, Hassan R, Rahmann Z, Haque KMF. Histopathological alterations induced by common krait bungarus caeruleus venom on hepatic, renal and cardiac tissues of albino mice. Int J Pharm Pharm Sci 2014;7:239-42.